484
Views
2
CrossRef citations to date
0
Altmetric
Review

Inflammatory myopathies: shedding light on promising agents and combination therapies in clinical trials

, &
Pages 1125-1140 | Received 04 Aug 2021, Accepted 04 Nov 2021, Published online: 12 Dec 2021

References

  • Meyer A, Meyer N, Schaeffer M, et al. Incidence and prevalence of inflammatory myopathies: a systematic review. Rheumatology (Oxford). 2015;54:50–63.
  • Schmidt J. Current classification and management of inflammatory myopathies. J Neuromuscul Dis. 2018;5:109–129.
  • Lilleker JB, Vencovsky J, Wang G, et al. The euroMyositis registry: an international collaborative tool to facilitate myositis research. Ann Rheum Dis. 2018;77:30–39.
  • Mohannak N, Pattison G, Hird K, et al. Dysphagia in patients with sporadic inclusion body myositis: management challenges. Int J Gen Med. 2019;12:465–474.
  • Aggarwal R, Rider LG, and Ruperto N, et al. 2016 American College of Rheumatology/European League against rheumatism criteria for minimal, moderate, and major clinical response in adult dermatomyositis and polymyositis: an international myositis assessment and clinical studies group/paediatric rheumatology international trials organisation collaborative initiative. Ann Rheum Dis. 2017;76:792–801
  • Montero-Melendez T. ACTH: the forgotten therapy. Semin Immunol. 2015;27:216–226.
  • Levine T. Treating refractory dermatomyositis or polymyositis with adrenocorticotropic hormone gel: a retrospective case series. Drug Des Devel Ther. 2012;6:133–139.
  • Aggarwal R, Marder G, Koontz DC, et al. Efficacy and safety of adrenocorticotropic hormone gel in refractory dermatomyositis and polymyositis. Ann Rheum Dis. 2018;77:720–727.
  • Ho-Mahler N, Turner B, Eaddy M, et al. Treatment with repository corticotropin injection in patients with rheumatoid arthritis, systemic lupus erythematosus, and dermatomyositis/polymyositis. Open Access Rheumatol. 2020;12:21–28.
  • Bunch TW. Prednisone and azathioprine for polymyositis: long-term followup. Arthritis Rheum. 1981;24:45–48.
  • Villalba L, Hicks JE, Adams EM, et al. Treatment of refractory myositis: a randomized crossover study of two new cytotoxic regimens. Arthritis Rheum. 1998;41:392–399.
  • Olivo Pallo PA, de Souza FHC, Miossi R, et al. Mycophenolate mofetil in patients with refractory systemic autoimmune myopathies: case series. Adv Rheumatol. 2018;58:34.
  • Danieli MG, Calcabrini L, Calabrese V, et al. Intravenous immunoglobulin as add on treatment with mycophenolate mofetil in severe myositis. Autoimmun Rev. 2009;9:124–127.
  • Boswell JS, Costner MI. Leflunomide as adjuvant treatment of dermatomyositis. J Am Acad Dermatol. 2008;58:403–406.
  • de Souza RC, de Souza FHC, Miossi R, et al. Efficacy and safety of leflunomide as an adjuvant drug in refractory dermatomyositis with primarily cutaneous activity. Clin Exp Rheumatol. 2017;35:1011–1013.
  • Takada K, Katada Y, Ito S, et al. Impact of adding tacrolimus to initial treatment of interstitial pneumonitis in polymyositis/dermatomyositis: a single-arm clinical trial. Rheumatology (Oxford). 2020;59:1084–1093.
  • Rotella K, Alvarez MR, and Saperstein Y, et al. Tacrolimus-induced remission in drug resistant inflammatory myopathy: a case series. Rheumatology (Sunnyvale). 2018;8:238
  • Sharma N, Putman MS, Vij R, et al. Myositis-associated interstitial lung disease: predictors of failure of conventional treatment and response to tacrolimus in a US cohort. J Rheumatol. 2017;44:1612–1618.
  • Go DJ, Park JK, Kang EH, et al. Survival benefit associated with early cyclosporine treatment for dermatomyositis-associated interstitial lung disease. Rheumatol Int. 2016;36:125–131.
  • Kurita T, Yasuda S, Oba K, et al. The efficacy of tacrolimus in patients with interstitial lung diseases complicated with polymyositis or dermatomyositis. Rheumatology (Oxford). 2015;54:39–44.
  • Vencovský J, Jarosová K, Machácek S, et al. Cyclosporine A versus methotrexate in the treatment of polymyositis and dermatomyositis. Scand J Rheumatol. 2000;29:95–102.
  • Ibrahim F, Choy E, Gordon P, et al. Second-line agents in myositis: 1-year factorial trial of additional immunosuppression in patients who have partially responded to steroids. Rheumatology (Oxford). 2015;54:1050–1055.
  • Ruperto N, Pistorio A, Oliveira S, et al. Prednisone versus prednisone plus ciclosporin versus prednisone plus methotrexate in new-onset juvenile dermatomyositis: a randomised trial. Lancet. 2016;387:671–678.
  • Anh-Tu Hoa S, and Hudson M. Critical review of the role of intravenous immunoglobulins in idiopathic inflammatory myopathies. Semin Arthritis Rheum. 2017;46:488–508.
  • Dalakas MC, Illa I, Dambrosia JM, et al. A controlled trial of high-dose intravenous immune globulin infusions as treatment for dermatomyositis. N Engl J Med. 1993;329:1993–2000.
  • Miyasaka N, Hara M, and Koike T, et al. Effects of intravenous immunoglobulin therapy in Japanese patients with polymyositis and dermatomyositis resistant to corticosteroids: a randomized double-blind placebo-controlled trial. Mod Rheumatol. 2012;22:382–393.
  • Lim J, Eftimov F, Verhamme C, et al. Intravenous immunoglobulins as first-line treatment in idiopathic inflammatory myopathies: a pilot study. Rheumatology (Oxford). 2021;60:1784–1792.
  • Aggarwal R, Charles-Schoeman C, Schessl J, et al. Prospective, double-blind, randomized, placebo-controlled phase III study evaluating efficacy and safety of octagam 10% in patients with dermatomyositis (“ProDERM Study”). Medicine (Baltimore). 2021;100:e23677.
  • Hachulla E, Benveniste O, Hamidou M, et al. High dose subcutaneous immunoglobulin for idiopathic inflammatory myopathies and dysimmune peripheral chronic neuropathies treatment: observational study of quality of life and tolerance. Int J Neurosci. 2017;127:516–523.
  • Tjärnlund A, Tang Q, and Wick C, et al. Abatacept in the treatment of adult dermatomyositis and polymyositis: a randomised, phase IIb treatment delayed-start trial. Ann Rheum Dis. 2018;77:55–62.
  • Fasano S, Gordon P, Hajji R, et al. Rituximab in the treatment of inflammatory myopathies: a review. Rheumatology (Oxford). 2017;56:26–36.
  • Oddis CV, Reed AM, and Aggarwal R, et al. Rituximab in the treatment of refractory adult and juvenile dermatomyositis and adult polymyositis: a randomized, placebo-phase trial. Arthritis Rheum. 2013;65:314–324.
  • Aggarwal R, Bandos A, Reed AM, et al. Predictors of clinical improvement in rituximab-treated refractory adult and juvenile dermatomyositis and adult polymyositis. Arthritis Rheumatol (Hoboken, NJ). 2014;66:740–749.
  • Aggarwal R, Loganathan P, Koontz D, et al. Cutaneous improvement in refractory adult and juvenile dermatomyositis after treatment with rituximab. Rheumatology (Oxford). 2017;56:247–254.
  • Zong M, Dorph C, Dastmalchi M, et al. Anakinra treatment in patients with refractory inflammatory myopathies and possible predictive response biomarkers: a mechanistic study with 12 months follow-up. Ann Rheum Dis. 2014;73:913–920.
  • Sugihara T, Okiyama N, Watanabe N, et al. Interleukin-1 and tumor necrosis factor α blockade treatment of experimental polymyositis in mice. Arthritis Rheum. 2012;64:2655–2662.
  • Okiyama N, Sugihara T, Iwakura Y, et al. Therapeutic effects of interleukin-6 blockade in a murine model of polymyositis that does not require interleukin-17A. Arthritis Rheum. 2009;60:2505–2512.
  • Kondo M, Murakawa Y, Matsumura T, et al. A case of overlap syndrome successfully treated with tocilizumab: a hopeful treatment strategy for refractory dermatomyositis? Rheumatology (Oxford). 2014;53:1907–1908.
  • Narazaki M, Hagihara K, Shima Y, et al. Therapeutic effect of tocilizumab on two patients with polymyositis. Rheumatology (Oxford). 2011;50:1344–1346.
  • Quesada-Masachs E, Caballero CM. Myositis as a rare complication after tocilizumab treatment. Pediatr Rheumatol Online J. 2014;12:345.
  • Salomonsson S, Lundberg IE. Cytokines in idiopathic inflammatory myopathies. Autoimmunity. 2006;39:177–190.
  • Efthimiou P, Schwartzman S, Kagen LJ. Possible role for tumour necrosis factor inhibitors in the treatment of resistant dermatomyositis and polymyositis: a retrospective study of eight patients. Ann Rheum Dis. 2006;65:1233–1236.
  • Anandacoomarasamy A, Howe G, Manolios N. Advanced refractory polymyositis responding to infliximab. Rheumatology (Oxford). 2005;44:562–563.
  • Dastmalchi M, Grundtman C, Alexanderson H, et al. A high incidence of disease flares in an open pilot study of infliximab in patients with refractory inflammatory myopathies. Ann Rheum Dis. 2008;67:1670–1677.
  • Iannone F, Scioscia C, Falappone PCF, et al. Use of etanercept in the treatment of dermatomyositis: a case series. J Rheumatol. 2006;33:1802–1804.
  • The Muscle Study Group. A randomized, pilot trial of etanercept in dermatomyositis. Ann Neurol. 2011;70:427–436.
  • Brunasso AMG, Aberer W, and Massone C. New onset of dermatomyositis/polymyositis during anti-TNF-α therapies: a systematic literature review. ScientificWorldJournal. 2014;2014:179180.
  • Schiffenbauer A, Garg M, Castro C, et al. A randomized, double-blind, placebo-controlled trial of infliximab in refractory polymyositis and dermatomyositis. Semin Arthritis Rheum. 2018;47:858–864.
  • Higgs BW, Zhu W, Morehouse C, et al. A phase 1b clinical trial evaluating sifalimumab, an anti-IFN-α monoclonal antibody, shows target neutralisation of a type I IFN signature in blood of dermatomyositis and polymyositis patients. Ann Rheum Dis. 2014;73:256–262.
  • Moghadam-Kia S, Charlton D, and Aggarwal R, et al. Management of refractory cutaneous dermatomyositis: potential role of Janus kinase inhibition with tofacitinib. Rheumatology (Oxford). 2019;58:1011–1015 .
  • Ladislau L, Suárez-Calvet X, Toquet S, Landon-Cardinal O, Amelin D, Depp M, et al. JAK inhibitor improves type I interferon induced damage: proof of concept in dermatomyositis. Brain. 2018;141:1609–1621.
  • Paik JJ, Casciola-Rosen L, and Shin JY, et al. Study of tofacitinib in refractory dermatomyositis: an open-label pilot study of ten patients. Arthritis Rheumatol (Hoboken, NJ). 2021;73:858–865.
  • Lucattelli M, Fineschi S, Selvi E, et al. Ajulemic acid exerts potent anti-fibrotic effect during the fibrogenic phase of bleomycin lung. Respir Res. 2016;17:49.
  • Gonzalez EG, Selvi E, Balistreri E, et al. Synthetic cannabinoid ajulemic acid exerts potent antifibrotic effects in experimental models of systemic sclerosis. Ann Rheum Dis. 2012;71:1545–1551.
  • Jm S, de Hoff LS, Sk S. Intravenous human immunoglobulin and/or methylprednisolone pulse therapies as a possible treat-to-target strategy in immune-mediated necrotizing myopathies. Rheumatol Int. 2019;39:1201–1212.
  • Mammen Andrew L, Eleni T. Intravenous Immune Globulin for Statin-Triggered Autoimmune Myopathy. N Engl J Med. 2015;373:1677–1680.
  • Valiyil R, Casciola-Rosen L, Hong G, et al. Rituximab therapy for myopathy associated with anti-signal recognition particle antibodies: a case series. Arthritis Care Res (Hoboken). 2010;62:1328–1334.
  • Casal-Dominguez M, Pinal-Fernandez I, and Huapaya J, et al. Efficacy and adverse effects of methotrexate compared with azathioprine in the antisynthetase syndrome. Clin Exp Rheumatol. 2019;37(5):858–861 .
  • Aggarwal R, Oddis CV, Goudeau D, et al. Autoantibody levels in myositis patients correlate with clinical response during B cell depletion with rituximab. Rheumatology (Oxford). 2016;55:991–999.
  • Leclair V, Galindo-Feria AS, Dastmalchi M, et al. Efficacy and safety of rituximab in anti-synthetase antibody positive and negative subjects with idiopathic inflammatory myopathy: a registry-based study. Rheumatology (Oxford). 2019;58:1214–1220.
  • Tsuchiya H, Tsuno H, Inoue M, et al. Mycophenolate mofetil therapy for rapidly progressive interstitial lung disease in a patient with clinically amyopathic dermatomyositis. Mod Rheumatol. 2014;24:694–696.
  • Fischer A, Brown KK, and Du Bois RM, et al. Mycophenolate mofetil improves lung function in connective tissue disease-associated interstitial lung disease. J Rheumatol. 2013;40:640–646
  • Witt LJ, Demchuk C, Curran JJ, et al. Benefit of adjunctive tacrolimus in connective tissue disease-interstitial lung disease. Pulm Pharmacol Ther. 2016;36:46–52.
  • Bae S, Charles-Schoeman C. Oral cyclophosphamide in treatment of patients with refractory idiopathic inflammatory myopathies: a retrospective observational study. Clin Rheumatol. 2018;37:2113–2123.
  • Ge Y, Peng Q, Zhang S, et al. Cyclophosphamide treatment for idiopathic inflammatory myopathies and related interstitial lung disease: a systematic review. Clin Rheumatol. 2015;34:99–105.
  • Allenbach Y, Guiguet M, Rigolet A, et al. Efficacy of rituximab in refractory inflammatory myopathies associated with anti- synthetase autoantibodies: an open-label, phase II trial. PLoS ONE. 2015;10:e0133702.
  • Langlois V, Gillibert A, Uzunhan Y, et al. Rituximab and cyclophosphamide in antisynthetase syndrome-related interstitial lung disease: an observational retrospective study. J Rheumatol. 2020;47:1678–1686.
  • Saunders P, Tsipouri V, Keir GJ, et al. Rituximab versus cyclophosphamide for the treatment of connective tissue disease-associated interstitial lung disease (RECITAL): study protocol for a randomised controlled trial. Trials. 2017;18:275.
  • Hisanaga J, Kotani T, and Fujiki Y, et al. Successful multi‐target therapy including rituximab and mycophenolate mofetil in anti‐melanoma differentiation‐associated gene 5 antibody‐positive rapidly progressive interstitial lung disease with clinically amyopathic dermatomyositis. Int J Rheum Dis. 2017;12:2182–2185.
  • Kurasawa K, Arai S, Namiki Y, et al. Tofacitinib for refractory interstitial lung diseases in anti-melanoma differentiation-associated 5 gene antibody-positive dermatomyositis. Rheumatology (Oxford). 2018;57:2114–2119.
  • Chen Z, Wang X, Ye S. Tofacitinib in amyopathic dermatomyositis-associated interstitial lung disease. N Engl J Med. 2019;381:291–293.
  • Tsuji H, Nakashima R, Hosono Y, et al. Multicenter prospective study of the efficacy and safety of combined immunosuppressive therapy with high-dose glucocorticoid, tacrolimus, and cyclophosphamide in interstitial lung diseases accompanied by anti-melanoma differentiation-associated gene 5-positive dermatomyositis. Arthritis Rheumatol. 2020;72:488–498.
  • Fredi M, Bartoli F, Cavazzana I, et al. Calcinosis in poly-dermatomyositis: clinical and laboratory predictors and treatment options. Clin Exp Rheumatol. 2017;35:303–308.
  • Jiang W, Yang H, Li S, et al. Clinical features, treatments and outcomes of calcinosis in adult patients with dermatomyositis: a single cohort study. Rheumatology (Oxford). 2021;60:2958–2962.
  • Dubos M, Ly K, and Martel C, et al. Is rituximab an effective treatment of refractory calcinosis? BMJ Case Rep. 2016;2016:bcr2015213179 .
  • Wendel S, Venhoff N, Frye BC, et al. Successful treatment of extensive calcifications and acute pulmonary involvement in dermatomyositis with the Janus-Kinase inhibitor tofacitinib - A report of two cases. J Autoimmun. 2019;100:131–136.
  • Xie F, Williams P, and Batchelor R, et al. Successful treatment of dermatomyositis and associated calcinosis with Adalimumab. Clin Exp Dermatol. 2020;45:945–949.
  • Ma JE, Ernste FC, Davis MDP, et al. Topical sodium thiosulfate for calcinosis cutis associated with autoimmune connective tissue diseases: the mayo clinic experience, 2012-2017. Clin Exp Dermatol. 2019;44:e189–e192.
  • Schmidt K, Schmidt J. Inclusion body myositis: advancements in diagnosis, pathomechanisms, and treatment. Curr Opin Rheumatol. 2017;29:632–638.
  • Badrising UA, Mlc M-S, Ferrari MD, et al. Comparison of weakness progression in inclusion body myositis during treatment with methotrexate or placebo. Ann Neurol. 2002;51:369–372.
  • Li J, Kim SG, Blenis J. Rapamycin: one drug, many effects. Cell Metab. 2014;19:373–379.
  • Benveniste O, Hogrel J-Y, and Belin L, et al. Sirolimus for treatment of patients with inclusion body myositis: a randomised, double-blind, placebo-controlled, proof-of-concept, phase 2b trial. Lancet Rheumatol. 2021;3:e40–e48.
  • Dalakas MC, Rakocevic G, Schmidt J, et al. Effect of Alemtuzumab (CAMPATH 1-H) in patients with inclusion-body myositis. Brain. 2009;132:1536–1544.
  • Schmidt K, Kleinschnitz K, Rakocevic G, et al. Molecular treatment effects of alemtuzumab in skeletal muscles of patients with IBM. BMC Neurol. 2016;16:48.
  • Sá J, Costelha J, Marinho A. Inclusion body myositis treated with Alemtuzumab. Eur J Case Rep Intern Med. 2019;6:1368.
  • Kosmidis ML, Alexopoulos H, Tzioufas AG, et al. The effect of anakinra, an IL1 receptor antagonist, in patients with sporadic inclusion body myositis (sIBM): a small pilot study. J Neurol Sci. 2013;334:123–125.
  • Kosmidis ML, Pikazis D, Vlachoyiannopoulos P, et al. Trial of canakinumab, an IL-1β receptor antagonist, in patients with inclusion body myositis. Neurol Neuroimmunol Neuroinflamm. 2019;6:e581.
  • Dalakas MC, Sonies B, Dambrosia J, et al. Treatment of inclusion-body myositis with IVIg: a double-blind, placebo-controlled study. Neurology. 1997;48:712–716.
  • Walter MC, Lochmüller H, Toepfer M, et al. High-dose immunoglobulin therapy in sporadic inclusion body myositis: a double-blind, placebo-controlled study. J Neurol. 2000;247:22–28.
  • Dalakas MC, Koffman B, Fujii M, et al. A controlled study of intravenous immunoglobulin combined with prednisone in the treatment of IBM. Neurology. 2001;56:323–327.
  • Cherin P, Delain J-C, Jaeger C, et al. Subcutaneous immunoglobulin use in inclusion body myositis: a review of 6 cases. Case Rep Neurol. 2015;7:227–232.
  • Kenya M, Ito H. Inclusion body myositis treated with intravenous immunoglobulin: a long-term study. Neurology. 2015;84:7047.
  • Rj B, Herbelin L, Jt K, et al. Pilot trial of etanercept in the treatment of inclusion-body myositis. Neurology. 2006;66:S123–4.
  • Ahmed M, Machado PM, Miller A, et al. Targeting protein homeostasis in sporadic inclusion body myositis. Sci Transl Med. 2016;8:331ra41.
  • Mendell JR, Sahenk Z, Al-Zaidy S, et al. follistatin gene therapy for sporadic inclusion body myositis improves functional outcomes. Mol Ther. 2017;25:870–879.
  • Amato AA, Sivakumar K, Goyal N, et al. Treatment of sporadic inclusion body myositis with bimagrumab. Neurology. 2014;83:2239–2246.
  • Hanna MG, Badrising UA, and Benveniste O, et al. Safety and efficacy of intravenous bimagrumab in inclusion body myositis (RESILIENT): a randomised, double-blind, placebo-controlled phase 2b trial. Lancet Neurol. 2019;18:834–844.
  • Amato AA, Hanna MG, Machado PM, et al. Efficacy and safety of bimagrumab in sporadic inclusion body myositis: long-term extension of RESILIENT. Neurology. 2021;96:e1595–e1607.
  • Sivakumar K, Cochrane TI, Sloth B, et al. Long-term safety and tolerability of bimagrumab (BYM338) in sporadic inclusion body myositis. Neurology. 2020;95:e1971–e1978.
  • Pasquali L, Longone P, Isidoro C, et al. Autophagy, lithium, and amyotrophic lateral sclerosis. Muscle Nerve. 2009;40:173–194.
  • Kitazawa M, Trinh DN, LaFerla FM. Inflammation induces tau pathology in inclusion body myositis model via glycogen synthase kinase-3 beta. Ann Neurol. 2008;64:15–24.
  • Dimachkie MM, Barohn RJ. Inclusion body myositis. Neurol Clin. 2014;32:629–646. vii
  • Rutkove SB, Parker RA, Nardin RA, et al. A pilot randomized trial of oxandrolone in inclusion body myositis. Neurology. 2002;58:1081–1087.
  • Her M, Kavanaugh A. Alterations in immune function with biologic therapies for autoimmune disease. J Allergy Clin Immunol. 2016;137:19–27.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.